Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
J Biol Chem ; 300(1): 105479, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37981210

RESUMEN

Autophagy is a degradative pathway that plays an important role in maintaining cellular homeostasis. Dysfunction of autophagy is associated with the progression of neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Although one of the typical features of brain aging is an accumulation of redox-active metals that eventually lead to neurodegeneration, a plausible link between trace metal-induced neurodegeneration and dysregulated autophagy has not been clearly determined. Here, we used a cupric chloride-induced neurodegeneration model in MN9D dopaminergic neuronal cells along with ultrastructural and biochemical analyses to demonstrate impaired autophagic flux with accompanying lysosomal dysfunction. We found that a surge of cytosolic calcium was involved in cupric chloride-induced dysregulated autophagy. Consequently, buffering of cytosolic calcium by calbindin-D28K overexpression or co-treatment with the calcium chelator BAPTA attenuated the cupric chloride-induced impairment in autophagic flux by ameliorating dysregulation of lysosomal function. Thus, these events allowed the rescue of cells from cupric chloride-induced neuronal death. These phenomena were largely confirmed in cupric chloride-treated primary cultures of cortical neurons. Taken together, these results suggest that abnormal accumulation of trace metal elements and a resultant surge of cytosolic calcium leads to neuronal death by impairing autophagic flux at the lysosomal level.


Asunto(s)
Autofagia , Calcio , Cobre , Neuronas Dopaminérgicas , Lisosomas , Autofagia/efectos de los fármacos , Autofagia/genética , Calcio/metabolismo , Cobre/farmacología , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/ultraestructura , Lisosomas/metabolismo , Animales , Ratones , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citosol/metabolismo
2.
Elife ; 102021 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-34965204

RESUMEN

Dopaminergic (DA) neurons exert profound influences on behavior including addiction. However, how DA axons communicate with target neurons and how those communications change with drug exposure remains poorly understood. We leverage cell type-specific labeling with large volume serial electron microscopy to detail DA connections in the nucleus accumbens (NAc) of the mouse (Mus musculus) before and after exposure to cocaine. We find that individual DA axons contain different varicosity types based on their vesicle contents. Spatially ordering along individual axons further suggests that varicosity types are non-randomly organized. DA axon varicosities rarely make specific synapses (<2%, 6/410), but instead are more likely to form spinule-like structures (15%, 61/410) with neighboring neurons. Days after a brief exposure to cocaine, DA axons were extensively branched relative to controls, formed blind-ended 'bulbs' filled with mitochondria, and were surrounded by elaborated glia. Finally, mitochondrial lengths increased by ~2.2 times relative to control only in DA axons and NAc spiny dendrites after cocaine exposure. We conclude that DA axonal transmission is unlikely to be mediated via classical synapses in the NAc and that the major locus of anatomical plasticity of DA circuits after exposure to cocaine are large-scale axonal re-arrangements with correlated changes in mitochondria.


Asunto(s)
Axones/efectos de los fármacos , Cocaína/farmacología , Conectoma , Neuronas Dopaminérgicas/efectos de los fármacos , Animales , Axones/ultraestructura , Neuronas Dopaminérgicas/ultraestructura , Ratones , Ratones Transgénicos , Microscopía Electrónica , Mitocondrias/ultraestructura , Núcleo Accumbens/efectos de los fármacos
3.
Mol Brain ; 14(1): 58, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33757554

RESUMEN

Mitochondrial structural changes are associated with the regulation of mitochondrial function, apoptosis, and neurodegenerative diseases. PRKN is known to be involved with various mechanisms of mitochondrial quality control including mitochondrial structural changes. Parkinson's disease (PD) with PRKN mutations is characterized by the preferential degeneration of dopaminergic neurons in the substantia nigra pars compacta, which has been suggested to result from the accumulation of damaged mitochondria. However, ultrastructural changes of mitochondria specifically in dopaminergic neurons derived from iPSC have rarely been analyzed. The main reason for this would be that the dopaminergic neurons cannot be distinguished directly among a mixture of iPSC-derived differentiated cells under electron microscopy. To selectively label dopaminergic neurons and analyze mitochondrial morphology at the ultrastructural level, we generated control and PRKN-mutated patient tyrosine hydroxylase reporter (TH-GFP) induced pluripotent stem cell (iPSC) lines. Correlative light-electron microscopy analysis and live cell imaging of GFP-expressing dopaminergic neurons indicated that iPSC-derived dopaminergic neurons had smaller and less functional mitochondria than those in non-dopaminergic neurons. Furthermore, the formation of spheroid-shaped mitochondria, which was induced in control dopaminergic neurons by a mitochondrial uncoupler, was inhibited in the PRKN-mutated dopaminergic neurons. These results indicate that our established TH-GFP iPSC lines are useful for characterizing mitochondrial morphology, such as spheroid-shaped mitochondria, in dopaminergic neurons among a mixture of various cell types. Our in vitro model would provide insights into the vulnerability of dopaminergic neurons and the processes leading to the preferential loss of dopaminergic neurons in patients with PRKN mutations.


Asunto(s)
Neuronas Dopaminérgicas/ultraestructura , Células Madre Pluripotentes Inducidas/citología , Mitocondrias/ultraestructura , Ubiquitina-Proteína Ligasas/genética , Secuencia de Bases , Sistemas CRISPR-Cas , Células Cultivadas , Edición Génica , Técnicas de Sustitución del Gen , Genes Reporteros , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Microscopía Electrónica/métodos , Microscopía Fluorescente , Neurogénesis , ARN Guía de Kinetoplastida/genética , Esferoides Celulares , Tirosina 3-Monooxigenasa/genética
4.
Nat Commun ; 12(1): 1807, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33753743

RESUMEN

Mitochondria-lysosome contacts are recently identified sites for mediating crosstalk between both organelles, but their role in normal and diseased human neurons remains unknown. In this study, we demonstrate that mitochondria-lysosome contacts can dynamically form in the soma, axons, and dendrites of human neurons, allowing for their bidirectional crosstalk. Parkinson's disease patient derived neurons harboring mutant GBA1 exhibited prolonged mitochondria-lysosome contacts due to defective modulation of the untethering protein TBC1D15, which mediates Rab7 GTP hydrolysis for contact untethering. This dysregulation was due to decreased GBA1 (ß-glucocerebrosidase (GCase)) lysosomal enzyme activity in patient derived neurons, and could be rescued by increasing enzyme activity with a GCase modulator. These defects resulted in disrupted mitochondrial distribution and function, and could be further rescued by TBC1D15 in Parkinson's patient derived GBA1-linked neurons. Together, our work demonstrates a potential role of mitochondria-lysosome contacts as an upstream regulator of mitochondrial function and dynamics in midbrain dopaminergic neurons in GBA1-linked Parkinson's disease.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Glucosilceramidasa/genética , Lisosomas/genética , Mitocondrias/genética , Mutación , Enfermedad de Parkinson/genética , Células Cultivadas , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/ultraestructura , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Glucosilceramidasa/metabolismo , Humanos , Hidrólisis , Lisosomas/metabolismo , Lisosomas/ultraestructura , Microscopía Confocal , Microscopía Electrónica de Transmisión , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Enfermedad de Parkinson/metabolismo , Imagen de Lapso de Tiempo/métodos , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
5.
Mol Brain ; 14(1): 54, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33726789

RESUMEN

TMEM132D is a human gene identified with multiple risk alleles for panic disorders, anxiety and major depressive disorders. Defining a conserved family of transmembrane proteins, TMEM132D and its homologs are still of unknown molecular functions. By generating loss-of-function mutants of the sole TMEM132 ortholog in C. elegans, we identify abnormal morphologic phenotypes in the dopaminergic PDE neurons. Using a yeast two-hybrid screen, we find that NAP1 directly interacts with the cytoplasmic domain of human TMEM132D, and mutations in C. elegans tmem-132 that disrupt interaction with NAP1 cause similar morphologic defects in the PDE neurons. NAP1 is a component of the WAVE regulatory complex (WRC) that controls F-actin cytoskeletal dynamics. Decreasing activity of WRC rescues the PDE defects in tmem-132 mutants, whereas gain-of-function of TMEM132D in mammalian cells inhibits WRC, leading to decreased abundance of select WRC components, impaired actin nucleation and cell motility. We propose that metazoan TMEM132 family proteins play evolutionarily conserved roles in regulating NAP1 protein homologs to restrict inappropriate WRC activity, cytoskeletal and morphologic changes in the cell.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Citoesqueleto/ultraestructura , Neuronas Dopaminérgicas/ultraestructura , Proteínas de la Membrana/metabolismo , Morfogénesis/genética , Neurogénesis/genética , Células Receptoras Sensoriales/ultraestructura , Actinas/metabolismo , Animales , Evolución Biológica , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Forma de la Célula , Secuencia Conservada , Neuronas Dopaminérgicas/metabolismo , Mutación con Ganancia de Función , Genes Reporteros , Células HEK293 , Humanos , Mutación con Pérdida de Función , Familia de Multigenes , Complejos Multiproteicos/fisiología , Trastorno de Pánico/genética , Dominios Proteicos , Mapeo de Interacción de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Células Receptoras Sensoriales/metabolismo , Técnicas del Sistema de Dos Híbridos
6.
Autophagy ; 17(4): 855-871, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32286126

RESUMEN

Macroautophagy/autophagy cytoplasmic quality control pathways are required during neural development and are critical for the maintenance of functional neuronal populations in the adult brain. Robust evidence now exists that declining neuronal autophagy pathways contribute to human neurodegenerative diseases, including Parkinson disease (PD). Reliable and relevant human neuronal model systems are therefore needed to understand the biology of disease-vulnerable neural populations, to decipher the underlying causes of neurodegenerative disease, and to develop assays to test therapeutic interventions in vitro. Human induced pluripotent stem cell (hiPSC) neural model systems can meet this demand: they provide a renewable source of material for differentiation into regional neuronal sub-types for functional assays; they can be expanded to provide a platform for screening, and they can potentially be optimized for transplantation/neurorestorative therapy. So far, however, hiPSC differentiation protocols for the generation of ventral midbrain dopaminergic neurons (mDANs) - the predominant neuronal sub-type afflicted in PD - have been somewhat restricted by poor efficiency and/or suitability for functional and/or imaging-based in vitro assays. Here, we describe a reliable, monolayer differentiation protocol for the rapid and reproducible production of high numbers of mDANs from hiPSC in a format that is amenable for autophagy/mitophagy research. We characterize these cells with respect to neuronal differentiation and macroautophagy capability and describe qualitative and quantitative assays for the study of autophagy and mitophagy in these important cells.Abbreviations: AA: ascorbic acid; ATG: autophagy-related; BDNF: brain derived neurotrophic factor; CCCP: carbonyl cyanide m-chlorophenylhydrazone; dbcAMP: dibutyryl cAMP; DAN: dopaminergic neuron; DAPI: 4',6-diamidino-2-phenylindole; DAPT: N-[N-(3,5-difluorophenacetyl)-L-alanyl]-sphenylglycine; DLG4/PSD95: discs large MAGUK scaffold protein 4; DMEM: Dulbecco's modified eagle's medium; EB: embryoid body; ECAR: extracellular acidification rate; EGF: epidermal growth factor; FACS: fluorescence-activated cell sorting; FCCP: arbonyl cyanide p-triflouromethoxyphenylhydrazone; FGF: fibroblast growth factor; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GDNF: glia cell derived neurotrophic factor; hiPSC: human induced pluripotent stem cell; LAMP2A: lysosomal associated membrane protein 2A; LT-R: LysoTracker Red; MAP1LC3: microtubule associated protein 1 light chain 3; mDAN: midbrain dopaminergic neuron; MEF: mouse embryonic fibroblast; MT-GR: MitoTracker Green; MT-R: MitoTracker Red; NAS2: normal SNCA2; NEM: neuroprogenitor expansion media; NR4A2/NURR1: nuclear receptor subfamily group A member 2; OA: oligomycin and antimycin A; OCR: oxygen consumption rate; PD: Parkinson disease; SHH: sonic hedgehog signaling molecule; SNCA/α-synuclein: synuclein alpha; TH: tyrosine hydroxylase; VTN: vitronectin.


Asunto(s)
Autofagia , Técnicas de Cultivo de Célula , Neuronas Dopaminérgicas/citología , Células Madre Pluripotentes Inducidas/citología , Mitofagia , Autofagia/efectos de los fármacos , Autofagia/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/ultraestructura , Regulación de la Expresión Génica/efectos de los fármacos , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/ultraestructura , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Mesencéfalo/citología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitofagia/efectos de los fármacos , Mitofagia/genética , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/genética , Piridinas/farmacología , Pirimidinas/farmacología , Factores de Tiempo
7.
Neurochem Res ; 45(12): 2939-2948, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33037975

RESUMEN

Methylmercury (MeHg) is a potent neurotoxicant. The mechanisms underlying MeHg-induced neurotoxicity are not fully understood. Several studies have shown that protein chaperones are involved in MeHg toxicity. The protein co-chaperone, stress inducible protein 1 (STI-1), has important functions in protein quality control of the chaperone pathway. In the current study, dopaminergic (DAergic) cephalic (CEP) neuronal morphology was evaluated in the Caenorhabditis elegans (C. elegans) sti-1 knockout strain. In the control OH7193 strain (dat-1::mCherry + ttx-3::mCherry), we characterized the morphology of CEP neurons by checking the presence of attached vesicles and unattached vesicles to the CEP dendrites. We showed that the attached vesicles were only present in adult stage worms; whereas they were absent in the younger L3 stage worms. In the sti-1 knockout strain, MeHg treatment significantly altered the structures of CEP dendrites with discontinuation of mCherry fluorescence and shrinkage of CEP soma, as compared to the control. 12 h post treatment on MeHg-free OP50-seeded plates, the discontinuation of mCherry fluorescence of CEP dendrites in worms treated with 0.05 or 0.5 µM MeHg returned to levels statistically indistinguishable from control, while in worms treated with 5 µM MeHg a higher percentage of discontinuation of mCherry fluorescence persisted. Despite this strong effect by 5 µM MeHg, CEP attached vesicles were increased upon 0.05 or 0.5 µM MeHg treatment, yet unaffected by 5 µM MeHg. The CEP attached vesicles of sti-1 knockout strain were significantly increased shortly after MeHg treatment, but were unaffected 48 h post treatment. In addition, there was a significant interactive effect of MeHg and sti-1 on the number of attached vesicles. Knock down sti-1 via RNAi did not alter the number of CEP attached vesicles. Taking together, our data suggests that the increased occurrence of attached vesicles in adult stage worms could initiate a substantial loss of membrane components of CEP dendrites following release of vesicles, leading to the discontinuation of mCherry fluorescence, and the formation of CEP attached vesicles could be regulated by sti-1 to remove cellular debris for detoxification.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/efectos de los fármacos , Dendritas/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Proteínas de Choque Térmico/genética , Compuestos de Metilmercurio/toxicidad , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Dendritas/patología , Dendritas/ultraestructura , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/ultraestructura , Técnicas de Inactivación de Genes , Cabeza/inervación , Masculino
8.
Sci Rep ; 10(1): 15485, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32968089

RESUMEN

Mutations in PRKN are the most common cause of early onset Parkinson's disease. Parkin is an E3 ubiquitin ligase, functioning in mitophagy. Mitochondrial abnormalities are present in PRKN mutant models. Patient derived neurons are a promising model in which to study pathogenic mechanisms and therapeutic targets. Here we generate induced neuronal progenitor cells from PRKN mutant patient fibroblasts with a high dopaminergic neuron yield. We reveal changing mitochondrial phenotypes as neurons undergo a metabolic switch during differentiation. Fibroblasts from 4 controls and 4 PRKN mutant patients were transformed into induced neuronal progenitor cells and subsequently differentiated into dopaminergic neurons. Mitochondrial morphology, function and mitophagy were evaluated using live cell fluorescent imaging, cellular ATP and reactive oxygen species production quantification. Direct conversion of control and PRKN mutant patient fibroblasts results in induced neuronal progenitor and their differentiation yields high percentage of dopaminergic neurons. We were able to observe changing mitochondrial phenotypes as neurons undergo a metabolic switch during differentiation. Our results show that when pre-neurons are glycolytic early in differentiation mitophagy is unimpaired by PRKN deficiency. However as neurons become oxidative phosphorylation dependent, mitophagy is severely impaired in the PRKN mutant patient neurons. These changes correlate with changes in mitochondrial function and morphology; resulting in lower neuron yield and altered neuronal morphology. Induced neuronal progenitor cell conversion can produce a high yield of dopaminergic neurons. The mitochondrial phenotype, including mitophagy status, is highly dependent on the metabolic status of the cell. Only when neurons are oxidative phosphorylation reliant the extent of mitochondrial abnormalities are identified. These data provide insight into cell specific effects of PRKN mutations, in particular in relation to mitophagy dependent disease phenotypes and provide avenues for alternative therapeutic approaches.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Mitofagia , Oxidación-Reducción , Ubiquitina-Proteína Ligasas/genética , Adulto , Muerte Celular , Cromanos/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/ultraestructura , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/genética , Mitocondrias/metabolismo , Imagen Óptica , Oxidación-Reducción/efectos de los fármacos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Ubiquitina-Proteína Ligasas/fisiología
9.
Sci Rep ; 10(1): 8912, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488042

RESUMEN

Nigrostriatal dopamine (DA) projections are anatomically organized along the dorsolateral-ventromedial axis, conveying long-term value signals to the striatum for shaping actions toward multiple future rewards. The present study examines whether the topographic organization of long-term value signals are observed upon activity of presumed DA neurons and presumed striatal projection neurons (phasically active neurons, PANs), as predicted based on anatomical literature. Our results indicate that DA neurons in the dorsolateral midbrain encode long-term value signals on a short timescale, while ventromedial midbrain DA neurons encode such signals on a relatively longer timescale. Activity of the PANs in the dorsal striatum is more heterogeneous for encoding long-term values, although significant differences in long-term value signals were observed between the caudate nucleus and putamen. These findings suggest that topographic DA signals for long-term values are not simply transferred to striatal neurons, possibly due to the contribution of other projections to the striatum.


Asunto(s)
Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/fisiología , Animales , Núcleo Caudado/fisiología , Conducta de Elección/fisiología , Cuerpo Estriado/anatomía & histología , Neuronas Dopaminérgicas/ultraestructura , Femenino , Macaca fuscata/anatomía & histología , Macaca fuscata/fisiología , Masculino , Vías Nerviosas/anatomía & histología , Vías Nerviosas/fisiología , Corteza Prefrontal/fisiología , Putamen/fisiología , Recompensa
10.
Sci Rep ; 10(1): 10278, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32581291

RESUMEN

Mutations in the PARK2 gene encoding parkin, an E3 ubiquitin ligase, are associated with autosomal recessive early-onset Parkinson's disease (PD). While parkin has been implicated in the regulation of mitophagy and proteasomal degradation, the precise mechanism leading to neurodegeneration in both sporadic and familial PD upon parkin loss-of-function remains unknown. Cultures of isogenic induced pluripotent stem cell (iPSC) lines with and without PARK2 knockout (KO) enable mechanistic studies of the effect of parkin deficiency in human dopaminergic neurons. We used such cells to investigate the impact of PARK2 KO on the lysosomal compartment and found a clear link between parkin deficiency and lysosomal alterations. PARK2 KO neurons exhibited a perturbed lysosomal morphology with enlarged electron-lucent lysosomes and an increased lysosomal content, which was exacerbated by mitochondrial stress and could be ameliorated by antioxidant treatment. We also found decreased lysosomal enzyme activity and autophagic perturbations, suggesting an impairment of the autophagy-lysosomal pathway in parkin-deficient cells. Interestingly, activity of the GBA-encoded enzyme, ß-glucocerebrosidase, was increased, suggesting the existence of a compensatory mechanism. In conclusion, our data provide a unique characterization of the morphology, content, and function of lysosomes in PARK2 KO neurons and reveal an important new connection between mitochondrial dysfunction and lysosomal dysregulation in PD pathogenesis.


Asunto(s)
Neuronas Dopaminérgicas/patología , Lisosomas/patología , Trastornos Parkinsonianos/patología , Ubiquitina-Proteína Ligasas/genética , Línea Celular , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/ultraestructura , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Pluripotentes Inducidas , Mutación con Pérdida de Función , Lisosomas/ultraestructura , Microscopía Electrónica de Transmisión , Trastornos Parkinsonianos/genética
11.
Theranostics ; 10(7): 3000-3021, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194851

RESUMEN

Rationale: Contactin-associated protein-like 4 (CNTNAP4) belongs to the neurexin superfamily and has critical functions in neurological development and synaptic function. Loss of CNTNAP4 in interneurons has been linked to autism, schizophrenia, and epilepsy. CNTNAP4 is also highly enriched in dopaminergic (DA) neurons in the substantia nigra (SN), however, few studies have investigated the role of CNTNAP4 in DA neurons, and whether CNTNAP4 deficiency in DA neurons contributes to Parkinson's disease (PD) remains unclear. Methods: Effects of CNTNAP4 knockdown or overexpression on the DA MN9D cell line were assessed via Western blotting, immunocytochemistry, and RNA sequencing. An in vivo animal model, including CNTNAP4 knockout mice and stereotaxic injections of adeno-associated viral short-hairpin RNA with the tyrosine-hydroxylase promotor to silence CNTNAP4 in the SN, as well as the resulting physiological/behavioral effects, were evaluated via behavioral tests, Western blotting, immunohistochemistry, and transmission electron microscopy. Enzyme-linked immunosorbent assays (ELISAs) were performed to examine the cerebrospinal fluid (CSF) and plasma CNTNAP4 concentrations in PD patients. Results: We demonstrated that CNTNAP4 knockdown induced mitophagy and increased α-synuclein expression in MN9D cells. CNTNAP4 knockdown in the SN induced PD-like increases in SN-specific α-synuclein expression, DA neuronal degeneration, and motor dysfunction in mice. In addition, CNTNAP4 knockdown in SN-DA neurons increased autophagosomes and reduced synaptic vesicles in the SN. Furthermore, CNTNAP4 knockout mice showed movement deficits, nigral DA degeneration, and increased autophagy, which were consistent with the SN-specific knockdown model. We also found that CSF and plasma CNTNAP4 expression was increased in PD patients; in particular, plasma CNTNAP4 was increased in male PD patients compared with controls or female PD patients. Conclusion: Our findings suggest that CNTNAP4 deficiency may initiate phenotypes relevant to PD, of which we elucidated some of the underlying mechanisms.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Proteínas de la Membrana/deficiencia , Proteínas del Tejido Nervioso/deficiencia , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/líquido cefalorraquídeo , Trastornos Parkinsonianos/metabolismo , Animales , Línea Celular , Neuronas Dopaminérgicas/química , Neuronas Dopaminérgicas/ultraestructura , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Potencial de la Membrana Mitocondrial , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitofagia , Proteínas del Tejido Nervioso/sangre , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Proteínas del Tejido Nervioso/fisiología , Trastornos Parkinsonianos/patología , Fenotipo , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Técnicas Estereotáxicas , Sustancia Negra/metabolismo , Sustancia Negra/patología , Sinapsinas/biosíntesis , Sinapsinas/genética , Transcriptoma , alfa-Sinucleína/biosíntesis , alfa-Sinucleína/genética
12.
Curr Protoc Neurosci ; 91(1): e88, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32049438

RESUMEN

Parkinson's disease (PD) is an age-related neurodegenerative disorder characterized by motor symptoms such as tremor, slowness of movement, rigidity, and postural instability, as well as non-motor features like sleep disturbances, loss of ability to smell, depression, constipation, and pain. Motor symptoms are caused by depletion of dopamine in the striatum due to the progressive loss of dopamine neurons in the substantia nigra pars compacta. Approximately 10% of PD cases are familial arising from genetic mutations in α-synuclein, LRRK2, DJ-1, PINK1, parkin, and several other proteins. The majority of PD cases are, however, idiopathic, i.e., having no clear etiology. PD is characterized by progressive accumulation of insoluble inclusions, known as Lewy bodies, mostly composed of α-synuclein and membrane components. The cause of PD is currently attributed to cellular proteostasis deregulation and mitochondrial dysfunction, which are likely interdependent. In addition, neuroinflammation is present in brains of PD patients, but whether it is the cause or consequence of neurodegeneration remains to be studied. Rodents do not develop PD or PD-like motor symptoms spontaneously; however, neurotoxins, genetic mutations, viral vector-mediated transgene expression and, recently, injections of misfolded α-synuclein have been successfully utilized to model certain aspects of the disease. Here, we critically review the advantages and drawbacks of rodent PD models and discuss approaches to advance pre-clinical PD research towards successful disease-modifying therapy. © 2020 The Authors.


Asunto(s)
Neurotoxinas/toxicidad , Trastornos Parkinsonianos , Animales , Cuerpo Estriado/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/ultraestructura , Evaluación Preclínica de Medicamentos/métodos , Predicción , Estudio de Asociación del Genoma Completo , Técnicas Histológicas , Humanos , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/genética , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Plaguicidas/toxicidad , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Ratas , Sustancia Negra/efectos de los fármacos , Sinucleinopatías/genética , Sinucleinopatías/patología , alfa-Sinucleína/biosíntesis , alfa-Sinucleína/genética
13.
J Neurosci ; 40(3): 557-568, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31776210

RESUMEN

Mitochondria are important sources of energy, but they are also the target of cellular stress, toxin exposure, and aging-related injury. Persistent accumulation of damaged mitochondria has been implicated in many neurodegenerative diseases. One highly conserved mechanism to clear damaged mitochondria involves the E3 ubiquitin ligase Parkin and PTEN-induced kinase 1 (PINK1), which cooperatively initiate the process called mitophagy that identifies and eliminates damaged mitochondria through the autophagosome and lysosome pathways. Parkin is a mostly cytosolic protein, but is rapidly recruited to damaged mitochondria and target them for mitophagy. Moreover, Parkin interactomes also involve signaling pathways and transcriptional machinery critical for survival and cell death. However, the mechanism that regulates Parkin protein level remains poorly understood. Here, we show that the loss of homeodomain interacting protein kinase 2 (HIPK2) in neurons and mouse embryonic fibroblasts (MEFs) has a broad protective effect from cell death induced by mitochondrial toxins. The mechanism by which Hipk2-/- neurons and MEFs are more resistant to mitochondrial toxins is in part due to the role of HIPK2 and its kinase activity in promoting Parkin degradation via the proteasome-mediated mechanism. The loss of HIPK2 leads to higher cytosolic Parkin protein levels at basal conditions and upon exposure to mitochondrial toxins, which protects mitochondria from toxin-induced damage. In addition, Hipk2-/- neurons and MEFs show increased expression of PGC-1α (peroxisome proliferator-activated receptor-γ coactivator 1), a Parkin downstream target that can provide additional benefits via transcriptional activation of mitochondrial genes. Together, these results reveal a previously unrecognized avenue to target HIPK2 in neuroprotection via the Parkin-mediated pathway.SIGNIFICANCE STATEMENT In this study, we provide evidence that homeodomain interacting protein kinase 2 (HIPK2) and its kinase activity promote Parkin degradation via the proteasome-mediated pathway. The loss of HIPK2 increases cytosolic and mitochondrial Parkin protein levels under basal conditions and upon exposure to mitochondrial toxins, which protect mitochondria from toxin-induced damage. In addition, Hipk2-/- neurons and mouse embryonic fibroblasts also show increased expression of PGC-1α (peroxisome proliferator-activated receptor-γ coactivator 1), a Parkin downstream target that can provide additional benefits via transcriptional activation of mitochondrial genes. These results indicate that targeting HIPK2 and its kinase activity can have neuroprotective effects by elevating Parkin protein levels.


Asunto(s)
Mitocondrias/efectos de los fármacos , Neuronas , Fármacos Neuroprotectores , Neurotoxinas/toxicidad , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Neuronas Dopaminérgicas/ultraestructura , Femenino , Fibroblastos/metabolismo , Regulación de la Expresión Génica/genética , Masculino , Potencial de la Membrana Mitocondrial/genética , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/ultraestructura , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Proteínas Quinasas/genética
14.
Commun Biol ; 2: 418, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31754648

RESUMEN

Alpha-synuclein (α-syn) is an abundant neuroprotein elevated in cocaine addicts, linked to drug craving, and recruited to axon terminals undergoing glutamatergic plasticity - a proposed mechanism for substance abuse. However, little is known about normal α-syn function or how it contributes to substance abuse. We show that α-syn is critical for preference of hedonic stimuli and the cognitive flexibility needed to change behavioral strategies, functions that are altered with substance abuse. Electron microscopic analysis reveals changes in α-syn targeting of ventral tegmental area axon terminals that is dependent upon the duration of cocaine exposure. The dynamic changes in presynaptic α-syn position it to control neurotransmission and fine-tune the complex afferent inputs to dopamine neurons, potentially altering functional dopamine output. Cocaine also increases postsynaptic α-syn where it is needed for normal ALIX function, multivesicular body formation, and cocaine-induced exosome release indicating potentially similar α-syn actions for vesicle release pre- and post-synaptically.


Asunto(s)
Trastornos Relacionados con Cocaína/etiología , Trastornos Relacionados con Cocaína/metabolismo , Cocaína/metabolismo , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/metabolismo , Mesencéfalo/fisiopatología , alfa-Sinucleína/metabolismo , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Neuronas Dopaminérgicas/ultraestructura , Espacio Extracelular/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Motivación , Actividad Motora , Recompensa , Transducción de Señal , alfa-Sinucleína/genética
15.
Neurobiol Dis ; 132: 104609, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31494284

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disorder whereby loss of midbrain dopaminergic neurons results in motor dysfunction. Transplantation of human induced pluripotent stem cells (iPSCs) into the brain of patients affected by PD is one of the therapeutic approaches that has gained interest to compensate for the degeneration of neurons and improve disease symptoms. However, only a part of transplanted cells can differentiate into mature neurons while the majority remains in undifferentiated state. Here we investigated whether human neuronal precursor cells (hNPCs) derived from iPSCs have an active role in α-synuclein (α-syn) pathology. Our findings demonstrate that α-syn fibrils are taken up by hNPCs and are preferentially localized in lysosomes where they can be degraded. However, α-syn fibrils are also transferred between hNPCs in a cell-to-cell contact dependent manner, and are found in tunneling nanotube (TNT)-like structures. Thus, NPCs can have a dual role in the progression of α-syn pathology, which should be considered in human transplants.


Asunto(s)
Comunicación Celular/fisiología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/ultraestructura , Endocitosis/fisiología , alfa-Sinucleína/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Lisosomas/metabolismo , Células-Madre Neurales/metabolismo
16.
Int J Mol Med ; 44(4): 1414-1424, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31364729

RESUMEN

The balance between glutamate (cortex and thalamus) and dopamine (substantia nigra) inputs on striatal neurons is of vital importance. Dopamine deficiency, which breaks this balance and leads to the domination of cortical glutamatergic inputs, plays an important role in Parkinson's disease (PD). However, the exact impact on striatal neurons has not been fully clarified. Thus, the present study aimed to characterize the influence of corticostriatal glutamatergic inputs on striatal neurons after decortication due to dopamine depletion in rats. 6­Hydroxydopamine was injected into the right medial forebrain bundle to induce dopamine depletion, and/or ibotenic acid into the primary motor cortex to induce decortication. Subsequently, the grip strength test and Morris water maze task indicated that decortication significantly shortened the hang time and the latency that had been increased in the rats subjected to dopamine depletion. Golgi staining and electron microscopy analysis showed that the total dendritic length and dendritic spine density of the striatal neurons were decreased in the dopamine­depleted rats, whereas decortication alleviated this damage. Immunohistochemistry analysis demonstrated that decortication decreased the number of caspase­3­positive neurons in the dopamine­depleted rats. Moreover, reverse transcription­quantitative PCR and western blot analyses showed that decortication offset the upregulation of caspase­3 at both the protein and mRNA levels in the dopamine­depleted rats. In conclusion, the present study demonstrated that a relative excess of cortical glutamate inputs had a substantial impact on the pathological processes of striatal neuron lesions in PD.


Asunto(s)
Corteza Cerebral/metabolismo , Decorticación Cerebral , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Animales , Conducta Animal , Biomarcadores , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/ultraestructura , Inmunohistoquímica , Aprendizaje por Laberinto , Fuerza Muscular , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Ratas
17.
Neurochem Int ; 129: 104482, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31170424

RESUMEN

Dopamine (DA) neuron projections to the striatum are functionally heterogeneous with diverse behavioral roles. We focus here on DA neuron projections to the nucleus accumbens (NAc) medial Shell, their distinct anatomical and functional connections, and discuss their role in motivated behavior. We first review rodent studies showing that a subpopulation of DA neurons in the medial ventral tegmental area (VTA) project to the NAc medial Shell. Using a combinatorial strategy, we show that the majority of DA neurons projecting to the NAc Shell express vesicular glutamate transporter 2 (VGLUT2) making them capable of glutamate co-transmission (DA-GLU neurons). In the NAc dorsal medial Shell, all of the DA neuron terminals arise from DA-GLU neurons, while in the lateral NAc Shell, DA neuron terminals arise from both DA-GLU neurons and DA-only neurons, without VGLUT2. DA-GLU neurons make excitatory connections to the three major cells types, spiny projection neurons, fast-spiking interneuron and cholinergic interneurons (ChIs). The strongest DA-GLU neuron excitatory connections are to ChIs. Photostimulation of DA-GLU neuron terminals in the slice drives ChIs to burst fire. Finally, we review studies that address specially the behavioral function of this subpopulation of DA neurons in extinction learning and latent inhibition. Taking into account findings from anatomical and functional connectome studies, we propose that DA-GLU neuron connections to ChIs in the medial Shell play a crucial role in switching behavioral responses under circumstances of altered cue-reinforcer contingencies.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Ácido Glutámico/fisiología , Núcleo Accumbens/citología , Potenciales de Acción , Animales , Mapeo Encefálico , Neuronas Colinérgicas/fisiología , Conectoma , Señales (Psicología) , Neuronas Dopaminérgicas/efectos de la radiación , Neuronas Dopaminérgicas/ultraestructura , Extinción Psicológica/fisiología , Miedo/fisiología , Humanos , Interneuronas/fisiología , Ratones , Terminaciones Nerviosas/fisiología , Terminaciones Nerviosas/efectos de la radiación , Proteínas del Tejido Nervioso/metabolismo , Vías Nerviosas/anatomía & histología , Núcleo Accumbens/fisiología , Refuerzo en Psicología , Área Tegmental Ventral/anatomía & histología , Área Tegmental Ventral/fisiología , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
18.
Biochem Biophys Res Commun ; 513(4): 1048-1054, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31010680

RESUMEN

Attention deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders and is characterized by impaired attention, hyperactivity, and impulsivity. While multiple etiologies are implicated in ADHD, its underlying mechanism(s) remain unclear. Although previous studies have suggested dysregulation of dopaminergic signals, mitochondria, and brain-derived neurotrophic factor (BDNF) in ADHD, few studies have reported these associations directly. Stem cells from human exfoliated deciduous teeth (SHED) can efficiently differentiate into dopaminergic neurons (DNs) and are thus a useful disease-specific cellular model for the study of neurodevelopmental disorders associated with DN dysfunction. This study aimed to elucidate the relationships between DNs, mitochondria, and BDNF in ADHD by analyzing DNs differentiated from SHED obtained from three boys with ADHD and comparing them to those from three typically developing boys. In the absence of exogenous BDNF in the cell culture media, DNs derived from boys with ADHD (ADHD-DNs) exhibited impaired neurite outgrowth and branching, decreased mitochondrial mass in neurites, and abnormal intracellular ATP levels. In addition, BDNF mRNA was significantly decreased in ADHD-DNs. Supplementation with BDNF, however, significantly improved neurite development and mitochondrial function in ADHD-DNs. These results suggest that ADHD-DNs may have impaired neurite development and mitochondrial function associated with insufficient production of BDNF, which may be improved by exogenous BDNF supplementation. Findings such as these, from patient-derived SHED, may contribute to the future development of treatment strategies for aberrant dopaminergic signaling, mitochondrial functioning, and BDNF levels implicated in ADHD pathogenesis.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/patología , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Pulpa Dental/patología , Neuronas Dopaminérgicas/patología , Neuritas/efectos de los fármacos , Células Madre/patología , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Estudios de Casos y Controles , Células Cultivadas , Niño , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/ultraestructura , Humanos , Masculino , Mitocondrias/patología , Neuritas/ultraestructura , Diente Primario
19.
Adv Biosyst ; 3(9): e1900091, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-32648650

RESUMEN

Wnt signaling is a key developmental pathway that regulates dopaminergic progenitor cell proliferation and differentiation during neuronal development. This makes Wnt signaling an important therapeutic target for neurodegenerative conditions such as Parkinson's disease. Wnt signaling can be modulated using peptides such as UM206, which bind to the Wnt receptor Frizzled. Previous work has demonstrated remote activation of the Wnt pathway through Frizzled using peptide-functionalized magnetic nanoparticles (MNPs) with magnetic field stimulation. Using this technology, Wnt signaling is remotely activated in the neuronal cell line SH-SY5Y, and the phenotypic response to stimulation is assessed. Results indicate ß-catenin translocalization and activation of TCF/LEF responsive transcription in response to MNP and magnetic fields, which result in dopaminergic marker expression when synergistically combined with differentiation factors retinoic acid and the phorbol ester phorbol 12-myristate 13-acetate. This approach is translated into ex vivo postnatal rat brain slices modeling the developing nigrostriatal pathway. Dopaminergic marker expression is maintained in MNP-labeled SH-SY5Y cells after injection and magnetic stimulation. These results demonstrate the translational value of remote control of signal transduction for controlling neuronal precursor cell behavior and highlight the potential applications for controlled cell differentiation as part of cell therapies for neurodegenerative disease.


Asunto(s)
Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Nanopartículas de Magnetita/química , Mecanotransducción Celular/genética , Vía de Señalización Wnt/genética , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/ultraestructura , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Campos Magnéticos , Mecanotransducción Celular/efectos de los fármacos , Microtomía , Oligopéptidos/química , Ratas , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Técnicas de Cultivo de Tejidos , Transfección , Tretinoina/farmacología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/genética , beta Catenina/metabolismo
20.
Neuron ; 101(1): 103-118.e5, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30503644

RESUMEN

Stromalin, a cohesin complex protein, was recently identified as a novel memory suppressor gene, but its mechanism remained unknown. Here, we show that Stromalin functions as a negative regulator of synaptic vesicle (SV) pool size in Drosophila neurons. Stromalin knockdown in dopamine neurons during a critical developmental period enhances learning and increases SV pool size without altering the number of dopamine neurons, their axons, or synapses. The developmental effect of Stromalin knockdown persists into adulthood, leading to strengthened synaptic connections and enhanced olfactory memory acquisition in adult flies. Correcting the SV content in dopamine neuron axon terminals by impairing anterograde SV trafficking motor protein Unc104/KIF1A rescues the enhanced-learning phenotype in Stromalin knockdown flies. Our results identify a new mechanism for memory suppression and reveal that the size of the SV pool is controlled genetically and independent from other aspects of neuron structure and function through Stromalin.


Asunto(s)
Proteínas de Drosophila/deficiencia , Memoria/fisiología , Proteínas Nucleares/deficiencia , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/ultraestructura , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Proteínas Nucleares/genética , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/ultraestructura , Vesículas Sinápticas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...